Bone marrow-derived progenitor cells contribute to the pulmonary vascular remodeling in hypoxia-induced pulmonary hypertension

2004 ◽  
Vol 10 (5) ◽  
pp. S191
Author(s):  
Satoh Kimio ◽  
Karibe Akihiko ◽  
Sakuma Masahito ◽  
Kagaya Yutaka ◽  
Watanabe Jun ◽  
...  
Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3455-3455 ◽  
Author(s):  
Jason M. Aliotta ◽  
Mandy Pereira ◽  
Mark Dooner ◽  
Michael DelTatto ◽  
Elaine Papa ◽  
...  

Abstract Rationale: We have demonstrated that whole, undifferentiated bone marrow (WBM) cells isolated from mice with monocrotaline (MCT)-induced pulmonary hypertension (PH) cause pulmonary hypertensive changes when transplanted into lethally-irradiated mice (Aliotta et al., 2014 ATS abstract). In addition, mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs), sub-cellular particles shed by activated cells, can reverse features of MCT-induced PH (Aliotta et al., 2015 ATS abstract). Endothelial progenitor cells (EPCs) have been implicated in the pathogenesis of human PH as more have been noted to be present in the circulation and remodeled pulmonary vasculature of these patients compared with healthy humans. We wished to determine if bone marrow-derived EPCs isolated from mice with MCT PH were the cells responsible for inducing PH in healthy mice and if so, whether exposure of these EPCs to MSC EVs prevented the development of PH. Methods: Sca-1+/ckit+/flk1+ cells (EPCs) and sca-1-/ckit-/flk1- cells (non-EPCs) were isolated from the bone marrow of MCT-injured and vehicle-injected mice. Cohorts of lethally-irradiated mice were transplanted with EPCs (2,000 EPCs + 300,000 helper WBM cells/recipient) or non-EPCs (200,000 non-EPCs + 200,000 helper WBM cells/recipient) from the bone marrow of MCT-injured and vehicle-injected mice. 28 days after transplantation, recipient mouse right ventricular (RV) hypertrophy was assessed by RV-to-left ventricle+septum (RV/LV+S) ratio (mg/g) and pulmonary vascular remodeling by blood vessel wall thickness-to-diameter (WT/D) ratio. In addition, WBM from MCT-injured and vehicle-injected mice was cultured with or without MSC EVs for 48 hours. EPCs (80-100 EPCs + 300,000 helper WBM cells/recipient) and non-EPCs (500,000-700,000 non- EPCs + 300,000 helper WBM cells/recipient) were separated by flow cytometry and infused into lethally-irradiated mice which were analyzed 28 days post-transplant. Results: RV/LV+S ratios of mice transplanted with EPCs from MCT-injured mice (0.256+/-0.142 mg/g) were similar to those of MCT-injured mice (0.274+/-0.211 mg/g, p=NS, n=5/cohort) but elevated compared to mice transplanted with EPCs and non-EPCs from vehicle-injected mice (0.126+/-0.111 and 0.121+/-0.134 mg/g, p<0.05, n=5/cohort) and mice transplanted with non-EPCs from MCT-injured mice (0.139+/-0.218 mg/g, n=5/cohort) . WT/D ratios of mice transplanted with EPCs from MCT-injured mice (0.145+/-0.122) were similar to those of MCT-injured mice (0.156+/-0.232, p=NS, n=5/cohort) but elevated compared to mice transplanted with EPCs and non-EPCs from vehicle-injected mice (0.076+/-0.008 and 0.072+/-0.009, p<0.05, n=5/cohort) and mice transplanted with non-EPCs from MCT-injured mice (0.081+/-0.012, n=5/cohort). Mice transplanted with EPCs from MCT-injured mice that were treated with MSC EVs prior to transplantation had significantly lower RV/LV+S and WT/D ratios compared with mice transplanted with EPCs from MCT-injured mice that were not treated with MSC EVs prior to transplantation (0.119+/-0.164 mg/g, 0.072+/-0.008 vs. 0.188+/-0.201 mg/g, 0.122+/-0.011 p<0.05, n=5/cohort). Conclusions: These findings suggest that EPCs are among the pathogenic bone marrow cells in MCT-injured mice that are responsible for inducing RV hypertrophy and pulmonary vascular remodeling, key features of PH, upon transplantation into healthy mice. MSC EVs appear to be capable of reversing this disease via interactions with EPCs. Disclosures No relevant conflicts of interest to declare.


CHEST Journal ◽  
2005 ◽  
Vol 127 (5) ◽  
pp. 1793-1798 ◽  
Author(s):  
Kentaro Hayashida ◽  
Jun Fujita ◽  
Yoshiko Miyake ◽  
Hiroshi Kawada ◽  
Kiyoshi Ando ◽  
...  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 5190-5190
Author(s):  
Jason M. Aliotta ◽  
Mandy Pereira ◽  
Ashley Amaral ◽  
Arina Sorokina ◽  
Sharon I Rounds ◽  
...  

Abstract Abstract 5190 Rationale: Circulating endothelial cell-derived microparticles (MP) are altered in pulmonary arterial hypertension (PAH) but whether they are biomarkers of cellular injury or participants in disease pathogenesis is unknown. We have shown that lung-derived MP induce marrow cells to express lung-specific mRNA, protein. Objectives: Determine if lung, plasma-derived MP (LMP, PMP) alter pulmonary vascular endothelial or marrow progenitor cell phenotype to induce pulmonary vascular remodeling in PAH. Methods and Main Results: LMP, PMP isolated from monocrotaline (MCT)- or vehicle-treated mice were injected into healthy mice. Right ventricular (RV) hypertrophy and pulmonary vascular remodeling were assessed by RV-to-body weight (RV/BW), blood vessel wall thickness-to-diameter (WT/D) ratios. RV/BW, WT/D ratios were elevated in MCT vs. vehicle-injected mice (1. 99+0. 09vs. 1. 04+0. 09mg/g; 0. 159+0. 002vs. 0. 062+0. 009%). MP from MCT and vehicle-treated mice were quantitatively similar; however, MCT-LMP had higher endothelial cell mRNA expression and higher expression of mRNAs of proteins known to be abnormally expressed in PAH vs. vehicle-LMP. RV/BW, WT/D ratios were higher in mice injected with MCT-MP vs. mice injected with vehicle-MP (1. 63+0. 09vs. 1. 08+0. 09mg/g; 0. 113+0. 02vs. 0. 056+0. 01%). Lineage-depleted bone marrow cells co-cultured with MCT-MP and bone marrow cells isolated from MCT-MP infused mice had higher endothelial progenitor cell gene expression vs. cells co-cultured with vehicle-MP or cells isolated from vehicle MP infused mice. Conclusions: MP from MCT-injured mice have increased expression of growth factors implicated in PAH pathogenesis and induce RV hypertrophy, pulmonary vascular remodeling in healthy mice. Circulating MP may contribute to the development of MCT-induced pulmonary hypertension by inducing a pathologic phenotype in pulmonary vascular endothelial and endothelial progenitor cells. Disclosures: No relevant conflicts of interest to declare.


2003 ◽  
Vol 285 (1) ◽  
pp. L199-L208 ◽  
Author(s):  
Jan Herget ◽  
Jana Novotná ◽  
Jana Bíbová ◽  
Viera Povýšilová ◽  
Marie Vaňková ◽  
...  

Chronic hypoxia induces lung vascular remodeling, which results in pulmonary hypertension. We hypothesized that a previously found increase in collagenolytic activity of matrix metalloproteinases during hypoxia promotes pulmonary vascular remodeling and hypertension. To test this hypothesis, we exposed rats to hypoxia (fraction of inspired oxygen = 0.1, 3 wk) and treated them with a metalloproteinase inhibitor, Batimastat (30 mg/kg body wt, daily ip injection). Hypoxia-induced increases in concentration of collagen breakdown products and in collagenolytic activity in pulmonary vessels were inhibited by Batimastat, attesting to the effectiveness of Batimastat administration. Batimastat markedly reduced hypoxic pulmonary hypertension: pulmonary arterial blood pressure was 32 ± 3 mmHg in hypoxic controls, 24 ± 1 mmHg in Batimastat-treated hypoxic rats, and 16 ± 1 mmHg in normoxic controls. Right ventricular hypertrophy and muscularization of peripheral lung vessels were also diminished. Batimastat had no influence on systemic arterial pressure or cardiac output and was without any effect in rats kept in normoxia. We conclude that stimulation of collagenolytic activity in chronic hypoxia is a substantial causative factor in the pathogenesis of pulmonary vascular remodeling and hypertension.


2015 ◽  
Vol 308 (3) ◽  
pp. L229-L252 ◽  
Author(s):  
Steven C. Pugliese ◽  
Jens M. Poth ◽  
Mehdi A. Fini ◽  
Andrea Olschewski ◽  
Karim C. El Kasmi ◽  
...  

Hypoxic pulmonary hypertension (PH) comprises a heterogeneous group of diseases sharing the common feature of chronic hypoxia-induced pulmonary vascular remodeling. The disease is usually characterized by mild to moderate pulmonary vascular remodeling that is largely thought to be reversible compared with the progressive irreversible disease seen in World Health Organization (WHO) group I disease. However, in these patients, the presence of PH significantly worsens morbidity and mortality. In addition, a small subset of patients with hypoxic PH develop “out-of-proportion” severe pulmonary hypertension characterized by pulmonary vascular remodeling that is irreversible and similar to that in WHO group I disease. In all cases of hypoxia-related vascular remodeling and PH, inflammation, particularly persistent inflammation, is thought to play a role. This review focuses on the effects of hypoxia on pulmonary vascular cells and the signaling pathways involved in the initiation and perpetuation of vascular inflammation, especially as they relate to vascular remodeling and transition to chronic irreversible PH. We hypothesize that the combination of hypoxia and local tissue factors/cytokines (“second hit”) antagonizes tissue homeostatic cellular interactions between mesenchymal cells (fibroblasts and/or smooth muscle cells) and macrophages and arrests these cells in an epigenetically locked and permanently activated proremodeling and proinflammatory phenotype. This aberrant cellular cross-talk between mesenchymal cells and macrophages promotes transition to chronic nonresolving inflammation and vascular remodeling, perpetuating PH. A better understanding of these signaling pathways may lead to the development of specific therapeutic targets, as none are currently available for WHO group III disease.


Sign in / Sign up

Export Citation Format

Share Document